ArticlePDF Available

C1P Attenuates Lipopolysaccharide-Induced Acute Lung Injury by Preventing NF- B Activation in Neutrophils

Authors:

Abstract and Figures

Recently, ceramide-1-phosphate (C1P) has been shown to modulate acute inflammatory events. Acute lung injury (Arnalich et al. 2000. Infect. Immun. 68: 1942-1945) is characterized by rapid alveolar injury, lung inflammation, induced cytokine production, neutrophil accumulation, and vascular leakage leading to lung edema. The aim of this study was to investigate the role of C1P during LPS-induced acute lung injury in mice. To evaluate the effect of C1P, we used a prophylactic and therapeutic LPS-induced ALI model in C57BL/6 male mice. Our studies revealed that intrapulmonary application of C1P before (prophylactic) or 24 h after (therapeutic) LPS instillation decreased neutrophil trafficking to the lung, proinflammatory cytokine levels in bronchoalveolar lavage, and alveolar capillary leakage. Mechanistically, C1P inhibited the LPS-triggered NF-κB levels in lung tissue in vivo. In addition, ex vivo experiments revealed that C1P also attenuates LPS-induced NF-κB phosphorylation and IL-8 production in human neutrophils. These results indicate C1P playing a role in dampening LPS-induced acute lung inflammation and suggest that C1P could be a valuable candidate for treatment of ALI.
Content may be subject to copyright.
of January 29, 2016.
This information is current as
Activation in Neutrophils BκAcute Lung Injury by Preventing NF-
C1P Attenuates Lipopolysaccharide-Induced
Gómez-Muñoz, Holger K. Eltzschig and Marco Idzko
Korcan Ayata, Madelon Hossfeld, Nicolas Ehrat, Antonio
Kristin Baudiß, Rodolfo de Paula Vieira, Sanja Cicko,
ol.1402681
http://www.jimmunol.org/content/early/2016/01/22/jimmun
published online 22 January 2016J Immunol
Subscriptions http://jimmunol.org/subscriptions is online at: The Journal of ImmunologyInformation about subscribing to
Permissions http://www.aai.org/ji/copyright.html
Submit copyright permission requests at:
Email Alerts http://jimmunol.org/cgi/alerts/etoc
Receive free email-alerts when new articles cite this article. Sign up at:
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Immunologists, Inc. All rights reserved.
Copyright © 2016 by The American Association of
9650 Rockville Pike, Bethesda, MD 20814-3994.
The American Association of Immunologists, Inc.,
is published twice each month byThe Journal of Immunology
at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from
The Journal of Immunology
C1P Attenuates Lipopolysaccharide-Induced Acute Lung
Injury by Preventing NF-kB Activation in Neutrophils
Kristin Baudiß,* Rodolfo de Paula Vieira,* Sanja Cicko,* Korcan Ayata,*
Madelon Hossfeld,* Nicolas Ehrat,* Antonio Go
´mez-Mun
˜oz,
Holger K. Eltzschig,
and
Marco Idzko*
Recently, ceramide-1-phosphate (C1P) has been shown to modulate acute inflammatory events. Acute lung injury (Arnalich et al. 2000.
Infect. Immun. 68: 1942–1945) is characterized by rapid alveolar injury, lung inflammation, induced cytokine production, neutrophil
accumulation, and vascular leakage leading to lung edema. The aim of this study was to investigate the role of C1P during LPS-
induced acute lung injury in mice. To evaluate the effect of C1P, we used a prophylactic and therapeutic LPS-induced ALI model in
C57BL/6 male mice. Our studies revealed that intrapulmonary application of C1P before (prophylactic) or 24 h after (therapeutic)
LPS instillation decreased neutrophil trafficking to the lung, proinflammatory cytokine levels in bronchoalveolar lavage, and alveolar
capillary leakage. Mechanistically, C1P inhibited the LPS-triggered NF-kB levels in lung tissue in vivo. In addition, ex vivo
experiments revealed that C1P also attenuates LPS-induced NF-kB phosphorylation and IL-8 production in human neutrophils.
These results indicate C1P playing a role in dampening LPS-induced acute lung inflammation and suggest that C1P could be a
valuable candidate for treatment of ALI. The Journal of Immunology, 2016, 196: 000–000.
Sphingolipids are structural molecules of eukaryotic cell
membranes. However, they act as second messenger
molecules in the regulation of cell homeostasis. Further-
more, sphingolipids were recently shown to contribute to the
regulation of inflammatory responses and could modify the de-
velopment and progression of human diseases (1, 2). A patho-
physiological role of sphingolipids is implicated in diabetes,
insulin resistance, neurodegenerative disorders, atherosclerosis,
and allergic airway inflammation (1–5). The key molecule in the
sphingolipid metabolism is ceramide, which regulates vital cel-
lular functions such as apoptosis, cell growth, and differentiation.
An important metabolite is ceramide-1-phosphate (C1P), which is
generated through direct phosphorylation of ceramide by ceramide
kinase (CerK) (2, 5, 6). Although CerK is the only enzyme de-
scribed for the generation of C1P so far, there is convincing evi-
dence for the existence of a CerK-independent metabolic pathway
(7). There are still detectable levels of C1P in CerK-deficient
animals (8) and in baculovirus-infected Sf9 cells treated with
CerK inhibitor (9). Notably, although C1P was identified .20 y
ago, some of its biological functions have only been revealed in
the last few years. C1P has been shown to act either as an intra-
cellular second messenger (10) or as an extracellular mediator
binding to a recently functionally identified, but still not cloned,
specific G protein–coupled receptor upon secretion to the extra-
cellular milieu. This specific C1P-coupled receptor was discov-
ered and described only on RAW 264.7 macrophages yet (7, 11). It
has been shown that C1P regulates cell proliferation and apoptosis
(1–3, 5–7, 10–12). In addition, there is increasing evidence that
C1P plays a role in inflammatory responses, because it stimulates
phagocytosis of neutrophils (8, 13), induces migration of macro-
phages (11, 14), as well as synthesis of eicosanoids (15), and
modulates cytokine production by macrophages and mast cells (1,
5, 16, 17). Initial studies demonstrated an increase of ceramide
concentration in apoptotic cells after stimulation of J774 macro-
phages with LPS (16). Moreover, an anti-inflammatory effect of
C1P was shown in LPS-stimulated HEK 293 cells and human
PBMCs (18).
Acute lung injury (ALI) (19) occurs in the setting of an acute
severe illness accompanied by systemic inflammation (20, 21) and
is characterized by diffuse parenchymal pulmonary inflammation
and edema (20–23). Intratracheal (i.t.) and i.p. instillation of LPS,
a bacterial cell wall component, to rodents is a well-accepted
and common experimental model for ALI from pulmonary and
extrapulmonary origin (24, 25) featuring key pathological com-
ponents of the disease such as profound neutrophilic lung re-
cruitment, vascular leakage/lung edema, and subsequent systemic
inflammation (20, 26). Unfortunately, despite marked efforts and
multiple therapeutic strategies, the mortality rate of patients suf-
fering from ALI remains high (21, 27–29).
In this study, we used an LPS-induced mouse model of ALI to
investigate the effect of C8-C1P and C16-C1P treatment in a
prophylactic and therapeutic setting. To adapt our observations to a
human situation, we treated LPS-stimulated human blood isolated
neutrophils with C1P in vitro. As a potential pathomechanistic link,
the modulation of the NF-kB pathway by C1P was studied.
*Department of Pneumology, COPD and Asthma Research Group, University
Hospital Freiburg, 79106 Freiburg, Germany;
Department of Biochemistry and
Molecular Biology, University of the Basque Country, 48080 Bilbao, Spain; and
Organ Protection Program, Department of Anesthesiology, University of Colorado
School of Medicine, Aurora, CO 80045
ORCIDs: 0000-0001-7043-6692 (K.A.); 0000-0002-9964-6367 (N.E.).
Received for publication October 29, 2014. Accepted for publication December 14,
2015.
This study was supported by a grant from the German Research Foundation, ID 7/8–1
(to M.I.), National Institutes of Health Grants R01 DK097075, R01-HL0921, R01-
DK083385, R01-HL098294, and POIHL114457-01, and a grant from the Crohn’s
and Colitis Foundation of America (to H.K.E.).
Address correspondence and reprint requests to Dr. Marco Idzko, University
Hospital Freiburg, Department of Pneumology, COPD and Asthma Research
Group, Killianstrasse 5, 79106 Freiburg, Germany. E-mail address: marco.
idzko@uniklinik-freiburg.de
Abbreviations used in this article: ALI, acute lung injury; BALF, bronchoalveolar
lavage fluid; CerK, ceramide kinase; C1P, ceramide-1-phosphate; i.t., intratracheal;
KC, keratinocyte-derived chemokine; Mmp-9,matrix metalloproteinase 9; qPCR,
quantitative PCR; Traf2, TNFR-associated factor 2.
Copyright Ó2016 by The American Association of Immunologists, Inc. 0022-1767/16/$30.00
www.jimmunol.org/cgi/doi/10.4049/jimmunol.1402681
Published January 22, 2016, doi:10.4049/jimmunol.1402681
at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from
Materials and Methods
Reagents
Two types of C1P differing in carbonyl-chain length were used in our
experiments. Natural C16-C1P was purchased from Matreya LLC (Pleasant
Gap, PA) and solubilized in sterile nanopure water on ice using a probe
sonicator as previously described (14). Synthetic C8-C1P was acquired
from Cayman Chemical (Ann Arbor, MI) in a 1% solution of C8-C1P in
ethanol. The solution was aliquoted and frozen at 220˚C. Cells and mice
were not affected through the maximum concentration of 0.0005% ethanol
99%. Vehicle control means equally diluted in ethanol concentration in PBS
or medium in comparison with the diluted ethanol concentration of C8-C1P.
Animal studies
All experiments were approved by the local animal ethics committee and
conducted according to the Helsinki convention for the use and care of
animals. Male C57BL/6 mice (6–8 wk old) were bred at the animal facility
of the University Hospital Freiburg under specific pathogen-free condi-
tions. All animal experiments were performed using five animals per group
and were repeated three times.
LPS model of acute lung inflammation. First, mice were anesthetized using
ketamine (10 mg/kg; Intervet; Bela-Pharm GmbH, Vechta, Germany)/
rompun (150 mg/kg; Bayer, Leverkusen, Germany) solution injected i.p.
Second, they received an i.t. injection of LPS (300 mg/kg; Escherichia coli
Serotype 026:B6; Sigma-Aldrich GmbH, Steinheim, Germany) diluted in
50 ml sterile PBS. In addition, indicated concentration of C1P or vehicle
diluted in 80 ml sterile PBS was administered i.t. 1 h before (prophylactic
study) or 24 h after (therapeutic study) the LPS application. Mice were
euthanized 24 h after the last i.t. application.
Bronchoalveolar lavage fluid analysis by flow cytometry. Lungs were
washed three times through i.t. cannulae with 1 ml PBS containing 0.5 mM
EDTA. Total cell number was evaluated and differential cell distribution
was determined by flow cytometry (FACSCalibur; BD Biosciences, San
Diego, CA), by using the software CellQuest version 3.3 (BD Biosciences)
and FlowJo version 10 (Tree Star, Ashland, OR) as previously described
(30). In brief, mouse bronchoalveolar lavage fluid (BALF) cells were in-
cubated with unlabeled anti-CD16/CD32 to block Fc receptors and stained
for 20 min with anti-CD11c allophycocyanin, anti–Ly-6G (Gr-1) FITC,
anti-CD3e PE-Cy7, anti-CD45R (B220) PE-Cy7, and anti-mouse F4/80
PE (all from eBioscience, Frankfurt, Germany), in PBS containing 0.5%
BSA and 0.1% sodium azide. Gating strategy was as follows: first, non-
erythrocytes have been gated using forward scatter and side scatter. Next,
a lymphocyte gate was defined based on FCS and CD3
+
/B220
+
PE-Cy7
fluorescence. Neutrophils were determined as Gr-1
+
, CD3
2
/B220
2
cells,
whereas macrophages were defined as F4/80
+
, CD11c
+
, and CD3
2
/B220
2
population. The percentages obtained for each cell type were multiplied by
the number of total BALF.
Cytokine analysis. The levels of IL-1b, IL-6, IL-8, MIP-2, keratinocyte-
derived chemokine (KC), IFN-g, and TNF-awere measured in BALF and
neutrophil cell culture supernatants using ELISAs (Duoset; R&D Systems,
Minneapolis, MN), according to the manufacturer’s instructions. The de-
tection limit was 2 pg/ml. Samples with values below the detection limit
were assigned 1 pg/ml as cytokine concentration.
Plasma leakage assay. Plasma vascular leakage was examined as previously
described (31). In brief, Evans blue dye conjugated to albumin (20 mg/kg)
was injected into the tail vein of mice. Thirty minutes later, the mice were
sacrificed and the lungs were perfused with PBS supplemented with 5 mM
EDTA. Perfused lungs were excised en bloc, dried, weighed, and snap
frozen in liquid nitrogen. The whole lung was homogenized in PBS
(1 ml/100 mg tissue) before incubation in formamide at 60˚C for 18 h.
The OD of the supernatant was determined spectrophotometrically at
620 nm after centrifugation at 5000 3gfor 30 min. The concentration of
the extravasated Evans blue in lung homogenate was calculated against
the standard curve, and the results were expressed as microgram of
Evans blue dye per gram lung tissue (31).
Histology. Frozen lung tissue sections were cut and stained with H&E. The
density of neutrophils in the lung parenchyma was obtained as previously
described (32–35). In brief, 20 photomicrographs of lung parenchyma
(excluding areas of pulmonary vessels) were randomly obtained at 3400
magnification. The area of the whole photograph and the area of light (air
area) were calculated using the Software Image Pro Plus 4.0 (Media Cy-
bernetics, Rockville, MD). By subtracting the air area from the total
photograph area, we obtained the parenchyma tissue area. Then the
number of neutrophils was counted in the tissue area according to the
morphological criteria. The results were depicted as number of neutrophils
per square millimeter of parenchymal tissue (neutrophils/mm
2
).
Immunofluorescence and neutrophil quantification. Four-micrometer-thick
frozen lung sections were placed on polysine slides (Thermo Scientific), air-
dried, and fixed with cold acetone for 10 min. Tissue sections were washed
with PBS and blocked 1 h at room temperature with 5% goat serum, 1%
BSA, 0.1% cold fish skin gelatin (Sigma-Aldrich), 0.1% Triton X-100, and
0.05% Tween 20 in TBS. Sections were incubated with 1:1000 diluted GR-1
Ab (clone RB6-8C5; BioLegend) for 1 h at room temperature. As secondary
Ab, goat anti-rat Alexa555 (Life Technologies) was used. DAPI was
added during the last 10 min of secondary Ab incubation. Fluoromount
mounting medium (Sigma) and coverslips (36) were used to finish the
preparation. For imaging, Axioplan2 microscope with 633oil immer-
sion objective, AxioCam, and HAL100 have been used (all from Zeiss).
For image acquisition and analysis, Axiovision software v4.9.1.0 (Zeiss)
was used. Twelve high-power vision fields per lung were used for
counting neutrophils.
Total and phosphorylated NF-kB expression in lung tissue. Lungs were
homogenized in radioimmunoprecipitation assay buffer, containing PMSF,
natrium orthovanadate, phosphatase inhibitor mixture B, and protease in-
hibitor mixture (Santa Cruz Biotechnology, Santa Cruz, CA) on ice for
15 min and centrifuged at 17,000 3gat 4˚C for 15 min to remove the cell
debris. The amount of proteins was quantified by Quick Start Bradford
Protein Assay (Bio-Rad Lab GmbH, Munich, Germany). For all samples,
50 mg protein was loaded in NuPAGE 4–12% Bis-Trigel (Invitrogen AG,
Carlsbad, CA) and transferred to nitrocellulose membrane. The membrane
was blocked with 5% milk powder and incubated with a primary Ab
against phospho–NF-kB p65 (1:500, rabbit monoclonal IgG; Cell Signal-
ing, Danvers, MA) overnight. Afterward we used HRP-conjugated sec-
ondary Abs against rabbit IgG (Cell Signaling) for 1 h. Primary Ab was
visualized and enhanced by chemiluminescence with SuperSignal West
Dura (Thermo Scientific, Rockford, IL). The amount of mouse b-actin
detected by monoclonal anti-actin Ab clone C4 served as loading control
(MP Biomedicals LLC, Solon, OH). The densitometric analysis was per-
formed using ImageJ.
RNA isolation, cDNA synthesis, and quantitative PCR. Total RNA was
isolated with QIAzol lysis reagent for gene expression analysis (QIAGEN
GmbH, Hilden, Germany) following the manufacturer’s protocol. cDNA
synthesis was carried out using the First Strand cDNA synthesis kit
(Thermo Fisher Scientific GmbH, Schwerte, Germany). quantitative PCR
(qPCR) was performed on a LightCycler 480 (Roche Diagnostic GmbH,
Mannheim, Germany) using qPCR SYBR Green mix (Thermo Fisher
Scientific GmbH). b
2
-MICROGLOBULIN and GAPDH served as reference
genes. For all reactions the annealing temperature was 60˚C. Primer design
and relative quantifications were done as previously described (37); primer
sequences are available upon request.
In vitro studies
Human neutrophil isolation and culture. Human blood neutrophils were
obtained from venous blood using human Pancoll gradient (PAN-Biotech
GmbH, Aidenbach, Germany) as previously described (38). Isolated neu-
trophils were resuspended in PBS and their purity determined by using
Giemsa staining (.98%). Neutrophils were seeded in 24-well plates (2 3
10
6
/well) in RPMI 1640 supplemented with 10% FCS and penicillin/
streptomycin, and incubated at 37˚C with 5% CO
2
in a humidified at-
mosphere. Neutrophils were stimulated with C1P (1, 10 mM) or vehicle
for1hbeforeand1hafterLPS(1.5mg/ml) stimulation. Finally, su-
pernatant was collected 6 h after the last treatment and analyzed for IL-8
concentration.
NF-kB phosphorylation assay. Human neutrophils were seeded in six-well
plates (5 310
6
/well) in RPMI 1640 with 0.1% FCS and penicillin/
streptomycin. One hour before a 15-min LPS stimulation (1.5 mg/ml),
cells were preincubated with C1P 1 mM. Subsequently, collected cells
were wash ed in cold PBS a nd pr oteins wer e ext rac ted using radio-
immunoprecipitation assay lysis buffer as described earlier. Equal amounts
of proteins (20 mg) were analyzed for b-Actin and phospho–NF-kB p65
(Ser
536
) (rabbit monoclonal IgG; Cell Signaling).
Statistical analysis. If not stated otherwise, groups were compared using
one-way ANOVA, followed by Bonferroni comparison test (GraphPad
Prism 5 Software, San Diego, CA). The pvalues ,0.05 were regarded
as significant.
Results
Effects of C1P on LPS-induced ALI
i.t. instillation of LPS (300 mg/kg) in C57BL/6 for 24 or 48 h
resulted in severe acute lung inflammation, as demonstrated by
increased BALF cell numbers (neutrophils and macrophages),
2 C1P REDUCES LPS-INDUCED ACUTE LUNG INJURY
at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from
elevated BALF-cytokine levels (Figs. 1, 2), plasma leakage into
the lungs (Fig. 3), and leukocyte infiltration in lung parenchyma
(Fig. 4). The prophylactic administration of both 1 and 10 mM
C1P, 1 h before LPS administration, resulted in lower numbers of
neutrophils and macrophages in BALF (Fig. 1A), decreased levels
of proinflammatory cytokines, notably KC and MIP-2 (Fig. 1B),
IL-6, and TNF-a(Fig. 1C), and significant reduction in micro-
vascular plasma leakage to the lung (Fig. 3A). This was accom-
panied by a decreased amount of neutrophils (Fig. 4A, 4E) and
inflammatory infiltrate in lung parenchyma (Fig. 4C).
Interestingly, there seems to be a dose response for C8-C1P, but
not for C16-C1P. Especially in the prophylactic LPS model, cy-
tokine levels of MIP-2 or IL-6 are more reduced by the treatment
of C8-C1P 10 mM instead of C8-C1P 1 mM. Administration of
C16-C1P independent of the concentration showed similar effects.
Notably, the beneficial effects of C1P on the cardinal features of
LPS-induced lung inflammation were also observed when animals
were treated with C1P 24 h after the administration of LPS-induced
ALI (Figs. 2, 3B). Histological examination of the lungs with H&E
and immunohistochemistry staining with Gr-1 for neutrophils from
mice treated with LPS for 48 h showed a significantly higher
number of neutrophils in the lung parenchyma than in the lungs of
C1P-treated mice (Fig. 4B, 4D, 4F), suggesting that C1P can not
only prevent but also treat established ALI. In addition, previous
experiments have shown that C1P reduced the early inflammatory
response after an LPS stimulation of 6 h (data not shown).
C1P reduced phosphorylation of NF-kB p65 expression in lung
tissue
LPS-induced lung injury has been reported to increase NF-kBac-
tivation in the lungs (39). Thus, we next questioned whether C1P
can decrease LPS-induced NF-kB activation in the prophylactic
and therapeutic settings of ALI. As shown in Fig. 5A, C16-C1P and
C8-C1P 1 mM significantly reduced LPS-induced NF-kB p65 ac-
tivation in the prophylactic model. Similar effects of C1P are shown
on LPS-induced phospho–NF-kB p65 in the therapeutic setting.
C1P attenuated mRNA NF-kB2 expression in lung tissue
Furthermore, qPCR analysis revealed that the protective role of C1P
can also be linked to the reduced mRNA expression of NF-kB2
(Fig. 6A, 6D) and NF-kBtarget genes including CD83 molecule
(CD83)andmatrix metalloproteinase 9 (Mmp-9) (Fig. 6A, 6D),
CCR5 and IL-6 (Fig. 6B, 6E), Myd88 (Fig. 6B), TNFR-associated
factor 2 (Traf2) (Fig. 6E), and Foxp3 (Fig. 6C, 6F). Myd88 plays a
key role in the innate and adaptive immune response and is included
in the activation of several proinflammatory genes. Especially
mRNA level of IL-6 was highly induced by LPS and attenuated
with prophylactic and/or therapeutic C1P treatment. Reduced IL-15
mRNA by C1P leads to lower STAT3 mRNA, which is involved in
many cellular processes such as cell growth and apoptosis (Fig. 6C,
6F). To conclude, C1P reduced the expression of LPS-induced
genes, which are involved in the regulation of NF-kB signaling.
Effects of C1P on LPS-induced NF-kB activation and IL-8
production in neutrophils
Neutrophil recruitment and activation play a pivotal role in the
pathophysiology of ALI; thus, we investigated the effects of C1P
on LPS-induced IL-8 secretion and NF-kB activation in purified
human blood neutrophils.
According to our in vivo study, we determined the prophylactic
and the therapeutic effect of C1P on the LPS-induced IL-8 pro-
duction. Thus, neutrophils were treated with C1P (1 or 10 mM)
either 1 h before or 1 h after LPS stimulation (1.5 mg/ml). Su-
pernatants were collected after an additional 7 h. As shown in
Fig. 7, C1P administration before (Fig. 7A) or after (Fig. 7B) LPS
stimulation significantly decreased LPS-induced IL-8 production.
For the latter, neutrophils (5 310
6
) were incubated with C1P or
vehicle 60 min before stimulation with LPS for an additional
15 min. Pretreatment of neutrophils with C1P before LPS pulsing
led to significant reduction in phospho–NF-kB p65, compared
with vehicle-treated, LPS-stimulated neutrophils, so that treatment
with C1P significantly reduced the NF-kB activation (Fig. 7C). In
Fig. 7D we present a representative Western blotting for phospho–
NF-kB and b-actin.
Discussion
Sphingolipids have been show n pr ev io usly to act a s pr oin-
flammatory or anti-inflammatory agents (18, 40). Although
the role of sphingosine-1-phosphate in LPS-induced lung inflam-
mation has been extensively studied (12, 31, 41), the influence of
FIGURE 1. Prophylactic administration of C1P (1 and 10 mM) before LPS administration significantly reduced ALI. C8-C1P (upper panels) and C16-C1P
(lower panels) (1 and 10 mM) were given 1 h before LPS administration and the animals were euthanized 24 h after LPS instillation. BALF cell differential
count was measured by flow cytometry (A). Concentration of KC, MIP-2 (B) and IL-6, TNF-ain BALF (C) were determined by ELISA. One representative
experiment out of three is shown. Values are given as mean 6SEM. n= 5 mice in each group. *p,0.05, **p,0.01, ***p,0.001 versus vehicle/LPS.
The Journal of Immunology 3
at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from
C1P in this process is not well-known. Moreover, the exact
pathogenesis and molecular mechanisms leading to ALI (19) are
still poorly understood (20, 21, 42, 43). Hence specific therapies
have not been identified yet, and the current management for ALI
is mainly supportive (20, 21, 43). Our present study concentrates
on C1P as an anti-inflammatory modulator of LPS-induced acute
lung inflammation. A low-dose LPS model was used to reproduce
important biomarkers of ALI, such as edema formation and
proinflammatory cytokines release, according to the recommen-
dations from the American Thoracic Society (44).
In this article, we demonstrate for the first time, to our
knowledge, that both natural C16-C1P and the synthetic C8-C1P
analog attenuate LPS-induced ALI in mice. Intrapulmonary ap-
plication of C1P, before or after LPS administration, reduces the
amount of neutrophils and the production of proinflammatory
cytokines, and enhances the vascular leakage in the lung. Mech-
anistically we provide evidence that C1P inhibits LPS-induced
NF-kB2 mRNA expression and NF-kB activation in lung tissue
in vivo and neutrophils in vitro, and reduces LPS-primed IL-8
production by neutrophils.
ALI is characterized by pulmonary inflammation resulting from
microvascular endothelial barrier failure followed by a rich protein
pulmonary edema accumulation (20, 21, 26, 45, 46). Especially the
number of neutrophils seems to play a key role in the severity of
ALI (47). The intrapulmonary administration of LPS in rodents
has been accepted as a clinically relevant model of ALI (20, 26).
i.t. exposure of mice to LPS results in a massive recruitment of
neutrophils to the lungs as seen by increased BALF-neutrophilia at
24 and 48 h after LPS administration. The intrapulmonary treatment
of mice with C1P significantly reduces BALF-neutrophilia. Fur-
thermore, histological examination of the lungs from mice treated
with LPS and immunohistochemistry staining with Gr-1 display a
higher amount of neutrophils in the lung parenchyma in comparison
with mice that received C1P in a prophylactic or therapeutic setting.
In addition, LPS-induced proinflammatory cytokines, such as KC,
MIP-2, IL-6, and TNF-a, which actively participate in the patho-
genesis of ALI (e.g., by contributing to inflammatory cell recruit-
ment, activation, and migration) (20, 26, 48, 49), were attenuated by
C1P. Our findings were supported by Jo
´zefowski et al. (16), who
described C1P as a negative regulator of TNF-aproduction by LPS
in macrophages. Finally, IL-8 and its two homologs, KC and MIP-2
in mice, are important for the recruitment and activation of neu-
trophils (20, 26, 50) in ALI. IL-8 levels in BALF correlate with the
severity and prognosis of the disease (20, 50). Thus, our observation
that C1P inhibits these major cytokines involved in the induction
and maintenance of LPS-induced ALI underlines the potency of this
compound.
Nevertheless, differences are visible between C8-C1P concen-
trations, although C16-C1P independent of the concentration is
more stable in the effect. One explanation could be the natural
origin of the C16-C1P compound. Further pharmacokinetic and
pharmacodynamics studies could help to fully understand how
those active compounds were adsorbed and distributed in the body
to their capability to influence the immune system.
FIGURE 2. Therapeutic administration of C1P (1 and 10 mM) after LPS administration significantly reduced ALI. C8-C1P (upper panels) and C16-C1P
(lower panels) (1 and 10 mM) were given 24 h after LPS administration. Animals were euthanized 48 h after LPS instillation. BALF cell differential count
was analyzed by flow cytometry (A). KC, MIP-2 (B), IL-6, and TNF-awere determined in BALF by ELISA (C). One representative experiment of three is
shown. Values are given as mean 6SEM. n= 5 mice in each group. *p,0.05, **p,0.01, ***p,0.001 versus vehicle/LPS 48 h.
FIGURE 3. Prophylactic and therapeutic C1P treatment reduced LPS-induced plasma leakage. C1P (1 mM) was administered 1 h before LPS or 24 h
after LPS instillation. In the prophylactic study (A), animals were euthanized 24 h and in the therapeutic study (B) 48 h after LPS administration. Plasma
leakage was determined spectrophotometrically 18 h after Evans blue dye albumin (20 mg/kg) was injected into the tail vein. Values are given as mean 6
SEM. n= 5 mice in each group. The experiment was performed twice. *p,0.05, **p,0.01 versus vehicle/LPS 24 h or versus LPS 48 h/vehicle.
4 C1P REDUCES LPS-INDUCED ACUTE LUNG INJURY
at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from
FIGURE 4. Effect of C1P on neutrophil recruitment in ALI. C8-C1P (upper panel) or C16-C1P treatment (lower panel) (1 and 10 mM) were given 1 h
before LPS or 24 h after LPS administration. For the prophylactic study (A), animals were euthanized 24 h after LPS and for therapeutic study 48 h after
LPS instillation (B). Histological pictures from stained lungs with H&E: prophylactic treatment (C) with C1P and therapeutic administration (D) of C1P.
Scale bars, 50 mm. Fluorescently labeled neutrophils in the lung parenchyma were counted: prophylactic treatment with C1P (1 mM) (E) and therapeutic
administration of C1P (1 mM) (F). Values are given as mean 6SEM. n= 5 mice in each group. *p,0.05, **p,0.01, ***p,0.001 versus vehicle/LPS
24 h or versus vehicle/LPS 48 h.
The Journal of Immunology 5
at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from
Furthermore, the anti-inflammatory effect of C1P seems strongly
influenced by the used C1P concentration. In our previous study,
we already described only an anti-inflammatory effect until a C1P
concentration of 10 mM (51), which is in line with the results of
Hankins et al. (18). Often a proinflammatory effect of C1P (14,
52) is described either with higher C1P concentrations or different
solvent (53, 54). Further studies could clarify the full potential of
the C1P concentration regarding the interaction with the cells and
the role as exogenous or endogenous C1P compound.
A cardinal feature of LPS-induced ALI is the presence of
vascular leakage, leading to the development of pulmonary edema
(20, 26). A significant improvement in lung edema was observed
by pretreatment and posttreatment of animals with C1P. The re-
duction in lung edema might be related to the inhibition of neu-
trophil recruitment, because neutrophils are considered as the
primary cellular effectors of alveolar-capillary damage in ALI.
Notably, the potent anti-inflammatory capacity of C1P in ALI is
further supported by the fact that this effect was still observed in
mice with established LPS-induced lung inflammation.
The pathogenesis of ALI is complex and implies various signal
transduction processes. Particular attention has been given to the
NF-kB pathway, which regulates the expression of genes encoding
FIGURE 5. Prophylactic (A) and therapeutic
(C) C1P administration reduced NF-kB p65
protein expression and activation in lung tissue.
C1P (1 mM) was given before or 24 h after LPS
administration. Animals were euthanized 24 or
48 h after LPS administration, respectively. The
densitometric analysis of phospho–NF-kΒp65
was performed using ImageJ. Western blotting
analysis of the total extract (50 mg) using
phospho–NF-kB p65 Ab. Western blot of the
NF-kB p65 phosphorylation (upper blot)is
shown (Band D). The protein loading control
was monitored by staining the same membrane
with b-actin (lower blot)(Band D). The black
lines indicate where parts of the image were
joined. Values are given as mean 6SEM. n=5
mice in each group. **p,0.05, ***p,0.001
versus vehicle/LPS 24 h or versus vehicle/LPS
48 h.
FIGURE 6. C1P attenuated NF-kBmRNA expression and NF-kB–related genes in lung tissue of mice. The effect of prophylactic (AC) and therapeutic
(DF) C1P (10 mM) treatment in LPS-induced acute lung inflammation on NF-kB2 and NF-kBtarget genes are shown. qPCR was performed for NF-kB2,
MMP-9, and CD83 molecule (CD83)(Aand D), CCR5 and IL-6 (Band E), Myd88 (B) and Traf2 (E), IL-15,STAT3, and Foxp3 (Cand F). b
2
-MICRO-
GLOBULIN and GAPDH were used as reference genes. Values are mean 6SEM. Values are means 6SEMs of at least two biological replicates. n= 4–5
mice in each group of two technical replicates. *p,0.05, **p,0.01, ***p,0.001 versus vehicle/LPS 24 h or versus LPS 48 h/vehicle.
6 C1P REDUCES LPS-INDUCED ACUTE LUNG INJURY
at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from
mediators involved in inflammatory lung process (55, 56). In-
creased nuclear translocation of NF-kB in the lungs of patients
with ALI has been reported to correlate with disease severity and
outcome (19, 36, 55–57). Moreover, inhibition of NF-kB activa-
tion leads to reduction of acute lung inflammation in experimental
models of ALI (48, 56), pointing out the crucial role of the NF-kB
pathway in the pathogenesis of ALI. Furthermore, increased NF-
kB activation in neutrophils of ALI patients showed an important
correlation with impaired outcomes of the disease, particularly with
diminished time in the ventilator postincubation and increased sur-
vival in critically ill patients with ALI (56). Interestingly, we ob-
served that C1P was able to attenuate LPS-evoked NF-kB2 mRNA
expression in the lung tissue of animals. In addition, we observed an
inhibitory effect of C1P on NF-kB–related genes such as CCR5,
Mmp9,CD83,Myd88,Traf2 ,IL-6,IL-15,STAT3,andFoxp3,which
are implied in the inflammatory process. Moreover, Western blot
assays of NF-kB phosphorylation (phospho–NF-kB p65) encourage
our finding that C1P affects the LPS-induced NF-kBsignaling
pathway in the lung of mice. Similarly, Hankins et al. (18) have
shown the inhibitory effect of C1P (10 mM) on LPS-triggered NF-
kB activation and cytokine release in human embryonic kidney cells.
Contrary to LPS stimulation, others reported that higher concentra-
tion of C1P (20 mM) activates NF-kB in the alveolar rat macrophage
cell line NR8383 (52) and in resting cells of the murine macrophage
cell line J774A.1 (14). These findings support the hypothesis of
Hankins et al. that the inhibition of NF-kB by exogenous C1P is
dose dependent and specific to TLR4.
Furthermore, to better clarify the role of C1P, cell culture ex-
periments with primary human neutrophils were performed. C1P
was able to inhibit LPS-triggered NF-kB p65 phosphorylation in
the prophylactic and the therapeutic model, which was also asso-
ciated with reduction in LPS-induced IL-8 production by these cells.
Although the role of S1P receptors in the pathogenesis of in-
flammatory disorders has been extensively studied (2, 4, 12), the
knowledge about a possible receptor of C1P and its interaction is
hardly described. C1P might exert its anti-inflammatory capacity
by suppressing the activation of the NF-kB pathway. We support
the argument of Hankins et al. (7, 18, 58) that fluctuations in C1P
levels determine its proinflammatory or anti-inflammatory effects,
and further studies are required to clarify the interaction of C1P
with the plasma membrane or possible delineated receptors, as
well as the effect of C1P in specific cell types. C1P can act either
as an intracellular second messenger (10) or as an extracellular
mediator binding to functionally identified, but still not cloned,
specific G protein–coupled receptor upon secretion to the extra-
cellular milieu (11). Further studies could focus especially on this
subject to fully understand the interaction of C1P and its molec-
ular mechanism.
Therefore, we conclude that C1P acts as an important anti-
inflammatory agent in LPS-induced acute lung inflammation.
Major characteristic parameters of ALI, such as neutrophil acti-
vation, proinflammatory cytokines, and vascular plasma leakage
are diminished. Mechanistically we showed that C1P attenuates
NF-kB activation in human neutrophils and the expression of NF-
kB–related genes in the lungs of mice. Thus, C1P and its analogs
offer novel therapeutic targets for the treatment of ALI.
Acknowledgments
We thank Zsofia Lazar for critically revising the manuscript. Further thanks
go to Jessica Beckert for assistance in mice experiments and cell culture.
Disclosures
The authors have no financial conflicts of interest.
References
1. Arana, L., P. Gangoiti, A. Ouro, M. Trueba, and A. Go
´mez-Mun
˜oz. 2010. Ceramide
and ceramide 1-phosphate in health and disease. Lipids Health Dis. 9: 15.
2. Chalfant, C. E., and S. Spiegel. 2005. Sphingosine 1-phosphate and ceramide
1-phosphate: expanding roles in cell signaling. J. Cell Sci. 118: 4605–4612.
3. Mitra, P., M. Maceyka, S. G. Payne, N. Lamour, S. Milstien, C. E. Chalfant, and
S. Spiegel. 2007. Ceramide kinase regulates growth and survival of A549 human
lung adenocarcinoma cells. FEBS Lett. 581: 735–740.
4. Idzko, M., H. Hammad, M. van Nimwegen, M. Kool, T. M
uller, T. Soullie
´,
M. A. M. Willart, D. Hijdra, H. C. Hoogsteden, and B. N. Lambrecht. 2006.
Local application of FTY720 to the lung abrogates experimental asthma by
altering dendritic cell function. J. Clin. Invest. 116: 2935–2944.
5. Lahiri, S., and A. H. Futerman. 2007. The metabolism and function of
sphingolipids and glycosphingolipids. Cell.Mol.LifeSci.64: 2270–2284.
6. Kihara, A., S. Mitsutake, Y. Mizutani, and Y. Igarashi. 2007. Metabolism
and biological functions of two phosphorylated sphingolipids, sphingosine
1-phosphate and ceramide 1-phosphate. Prog. Lipid Res. 46: 126–144.
FIGURE 7. C1P administration reduced IL-8 release and NF-kB expression/activity in human neutrophils. Blood neutrophils (2 310
6
/well) were
stimulated with C1P (10 mM) 1 h before (A) and 1 h after (B) LPS stimulation (1.5 mg/ml) for 7 h. Then the medium was collected for analysis of IL-8
levels by ELISA (Aand B). For Western blot analysis, C1P (10 mM) was administered for 60 min to human neutrophils. Afterward, 5 310
6
cells were
stimulated with LPS 15 min to evaluate NF-kB p65 phosphorylation by Western blotting. The densitometric analysis (C) was performed using ImageJ. *p,
0.05 versus vehicle/LPS. Representative blotting of neutrophils stimulated with C1P (10 mM) and LPS are shown in (D). The protein loading control was
monitored by staining the same membrane with b-actin (lower panel). Values are given as mean 6SEM. **p,0.01, ***p,0.001 versus vehicle/LPS or
LPS/vehicle.
The Journal of Immunology 7
at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from
7. Granado, M. H., P. Gangoiti, A. Ouro, L. Arana, M. Gonza
´lez, M. Trueba, and
A. Go
´mez-Mun
˜oz. 2009. Ceramide 1-phosphate (C1P) promotes cell migration
involvement of a specific C1P receptor. Cell. Signal. 21: 405–412.
8. Graf, C., B. Zemann, P. Rovina, N. Urtz, A. Schanzer, R. Reuschel,
D. Mechtcheriakova, M. M
uller, E. Fischer, C. Reichel, et al. 2008. Neutropenia
with impaired immune response to Streptococcus pneumoniae in ceramide
kinase-deficient mice. J. Immunol. 180: 3457–3466.
9. Graf, C., M. Klumpp, M. Habig, P. Rovina, A. Billich, T. Baumruker,
B. Oberhauser, and F. Bornancin. 2008. Targeting ceramide metabolism with a
potent and specific ceramide kinase inhibitor. Mol. Pharmacol. 74: 925–932.
10. Boath, A., C. Graf, E. Lidome, T. Ullrich, P. Nussbaumer, and F. Bornancin.
2008. Regulation and traffic of ceramide 1-phosphate produced by ceramide
kinase: comparative analysis to glucosylceramide and sphingomyelin. J. Biol.
Chem. 283: 8517–8526.
11. Levi,M.,M.M.Meijler,A.Go
´mez-Mun
˜oz, and T. Zor. 2010. Distinct receptor-
mediated activities in macrophages for natural ceramide-1-phosphate (C1P) and for
phospho-ceramide anal ogue-1 (PCERA-1). Mol. Cell. Endocrinol. 314: 248–255.
12. Fyrst, H., and J. D. Saba. 2010. An update on sphingosine-1-phosphate and other
sphingolipid mediators. Nat. Chem. Biol. 6: 489–497.
13. Hinkovska-Galcheva, V. T., L. A. Boxer, P. J. Mansfield, D. Harsh,
A. Blackwood, and J. A. Shayman. 1998. The formation of ceramide-1-
phosphate during neutrophil phagocytosis and its role in liposome fusion. J.
Biol. Chem. 273: 33203–33209.
14. Arana, L., M. Ordon
˜ez, A. Ouro, I.-G. Rivera, P. Gangoiti, M. Trueba, and A. Gomez-
Mun
˜oz. 2013. Ceramide 1-phosphate induces macrophage chemoattractant protein-1
release: involvement in ceramide 1-phosphate-stimulated cell migration. Am. J.
Physiol. Endocrinol. Metab. 304: E1213–E1226.
15. Lamour, N. F., P. Subramanian, D. S. Wijesinghe, R. V. Stahelin, J. V. Bonventre,
and C. E. Chalfant. 2009. Ceramide 1-phosphate is required for the translocation
of group IVA cytosolic phospholipase A2 and prostaglandin synthesis. J. Biol.
Chem. 284: 26897–26907.
16. Jo
´zefowski, S., M. Czerkies, A. Łukasik, A. Bielawska, J. Bielawski,
K. Kwiatkowska, and A. Sobota. 2010. Ceramide and ceramide 1-phosphate are
negative regulators of TNF-aproduction induced by lipopolysaccharide. J.
Immunol. 185: 6960–6973.
17. Mitsutake, S., T.-J. Kim, Y. Inagaki, M. Kato, T. Yamashita, and Y. Igarashi.
2004. Ceramide kinase is a mediator of calcium-dependent degranulation in mast
cells. J. Biol. Chem. 279: 17570–17577.
18. Hankins, J. L., T. E. Fox, B. M. Barth, K. A. Unrath, and M. Kester. 2011.
Exogenous ceramide-1-phosphate reduces lipopolysaccharide (LPS)-mediated
cytokine expression. J. Biol. Chem. 286: 44357–44366.
19.Arnalich,F.,E.Garcia-Palomero,J.Lo
´pez, M. Jime
´nez, R. Madero, J. Renart,
J. J. Va
´zquez, and C. Montiel. 2000. Predictive value of nuclear factor kappaB activity
and plasma cytokine levels in patients with sepsis. Infect. Immun. 68: 1942–1945.
20. Matute-Bello, G., C. W. Frevert, and T. R. Martin. 2008. Animal models of acute
lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 295: L379–L399.
21. Ware, L. B., and M. A. Matthay. 2000. The acute respiratory distress syndrome.
N. Engl. J. Med. 342: 1334–1349.
22. Matthay, M. A., and G. A. Zimmerman. 2005. Acute lung injury and the acute
respiratory distress syndrome: four decades of inquiry into pathogenesis and
rational management. Am. J. Respir. Cell Mol. Biol. 33: 319–327.
23. Rosseau, S., P. Hammerl, U. Maus, H. D. Walmrath, H. Sch
utte, F. Grimminger,
W. Seeger, and J. Lohmeyer. 2000. Phenotypic characterization of alveolar
monocyte recruitment in acute respiratory distress syndrome. Am. J. Physiol.
Lung Cell. Mol. Physiol. 279: L25–L35.
24. Kitamura, Y., S. Hashimoto, N. Mizuta, A. Kobayashi, K. Kooguchi, I. Fujiwara,
and H. Nakajima. 2001. Fas/FasL-dependent apoptosis of alveolar cells after
lipopolysaccharide-induced lung injury in mice. Am. J. Respir. Crit. Care Med.
163: 762–769.
25. Wu, Y., M. Singer, F. Thouron, M. Alaoui-El-Azher, and L. Touqui. 2002. Effect
of surfactant on pulmonary expression of type IIA PLA(2) in an animal model of
acute lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 282: L743–L750.
26. Eckle, T., A. Grenz, S. Laucher, and H. K. Eltzschig. 2008. A2B adenosine
receptor signaling attenuates acute lung injury by enhancing alveolar fluid
clearance in mice. J. Clin. Invest. 118: 3301–3315.
27. Tiruvoipati, R., J. Botha, and G. Peek. 2012. Effectiveness of extracorporeal
membrane oxygenation when conventional ventilation fails: valuable option or
vague remedy? J. Crit. Care 27: 192–198.
28. Villar, J., D. Sulemanji, and R. M. Kacmarek. 2014. The acute respiratory distress
syndrome: incidence and mortality, has it changed? Curr. Opin. Crit. Care 20: 3–9.
29. Wang, C. Y., C. S. Calfee, D. W. Paul, D. R. Janz, A. K. May, H. Zhuo,
G. R. Bernard, M. A. Matthay, L. B. Ware, and K. N. Kangelaris. 2014. One-year
mortality and predictors of death among hospital survivors of acute respiratory
distress syndrome. Intensive Care Med. 40: 388–396.
30. Lucattelli, M., S. Cicko, T. M
uller, M. Lommatzsch, G. De Cunto, S. Cardini,
W. Sundas, M. Grimm, R. Zeiser, T. D
urk, et al. 2011. P2X7 receptor signaling
in the pathogenesis of smoke-induced lung inflammation and emphysema. Am. J.
Respir. Cell Mol. Biol. 44: 423–429.
31. Peng, X., P. M. Hassoun, S. Sammani, B. J. McVerry, M. J. Burne, H. Rabb,
D. Pearse, R. M. Tuder, and J. G. N. Garcia. 2004. Protective effects of sphin-
gosine 1-phosphate in murine endotoxin-induced inflammatory lung injury. Am.
J. Respir. Crit. Care Med. 169: 1245–1251.
32. Vieira, R. P., V. F. de Andrade, A. C. S. Duarte, A. B. G. Dos Santos, T. Mauad,
M. A. Martins, M. Dolhnikoff, and C. R. F. Carvalho. 2008. Aerobic condi-
tioning and allergic pulmonary inflammation in mice. II. Effects on lung vascular
and parenchymal inflammation and remodeling. Am. J. Physiol. Lung Cell. Mol.
Physiol. 295: L670–L679.
33. Ramos, D. S., C. R. Olivo, F. D. T. Quirino Santos Lopes, A. C. Toledo,
M. A. Martins, R. A. Lazo Oso
´rio, M. Dolhnikoff, W. Ribeiro, and R. P. Vieira.
2010. Low-intensity swimming training partially inhibits lipopolysaccharide-
induced acute lung injury. Med. Sci. Sports Exerc. 42: 113–119.
34. Reis Gonc¸alves, C. T., C. G. Reis Gonc¸alves,F.M.deAlmeida,F.D.Lopes,
A.C.dosSantosDura
˜o, F. A. dos Santos, L. F. da Silva, T. Marcourakis,
H. C. Castro-Faria-Neto, R. P. Vieira, andM. Dolhnikoff. 2012. Protective effects of
aerobic exercise on acute lung injury induced by LPS in mice. Crit. Care 16: R199.
35. Vieira, R. P., A. C. Toledo, L. B. Silva, F. M. Almeida, N. R. Damaceno-
Rodrigues, E. G. Caldini, A. B. G. Santos, D. H. Rivero, D. C. Hizume,
F. D. T. Q. S. Lopes, et al. 2012. Anti-inflammatory effects of aerobic exercise in
mice exposed to air pollution. Med. Sci. Sports Exerc. 44: 1227–1234.
36. Bo
¨hrer, H., F. Qiu, T. Zimmermann, Y. Zhang, T. Jllmer, D. Ma
¨nnel,
B. W. Bo
¨ttiger, D. M. Stern, R. Waldherr, H. D. Saeger, et al. 1997. Role of
NFkappaB in the mortality of sepsis. J. Clin. Invest. 100: 972–985.
37. Jeong,H.-J.,H.-S.Chung,B.-R.Lee,S.-J.Kim,S.-J.Yoo,S.-H.Hong,andH.-M.Kim.
2003. Expression of proinflammatory cytokines via HIF-1alpha and NF-kappaB
activation on desferrioxamine-stimulated HMC-1 cells. Biochem. Biophys. Res.
Commun. 306: 805–811.
38. Petrache, I., V. Natarajan, L. Zhen, T. R. Medler, A. T. Richter, C. Cho,
W. C. Hubbard, E. V. Berdyshev, and R. M. Tuder. 2005. Ceramide upregulation
causes pulmonary cell apoptosis and emphysema-like disease in mice. Nat. Med.
11: 491–498.
39. Wang, F., Z.-F. Xia, X.-L. Chen, Y.-T. Jia, Y.-J. Wang, and B. Ma. 2009. An-
giotensin II type-1 receptor antagonist attenuates LPS-induced acute lung injury.
Cytokine 48: 246–253.
40. Gomez-Mun
˜oz, A., P. Gangoiti, L. Arana, A. Ouro, I.-G. Rivera, M. Ordon
˜ez,
and M. Trueba. 2013. New insights on the role of ceramide 1-phosphate in in-
flammation. Biochim. Biophys. Acta 1831: 1060–1066.
41. Szczepaniak, W. S., Y. Zhang, S. Hagerty, M. T. Crow, P. Kesari, J. G. N. Garcia,
A. M. K. Choi, B. A. Simon, and B. J. McVerry. 2008. Sphingosine 1-phosphate
rescues canine LPS-induced acute lung injury and alters systemic inflammatory
cytokine production in vivo. Transl. Res. 152: 213–224.
42. Matthay, M. A., L. B. Ware, and G. A. Zimmerman. 2012. The acute respiratory
distress syndrome. J. Clin. Invest. 122: 2731–2740.
43. Fro
¨hlich, S., N. Murphy, D. Ryan, and J. F. Boylan. 2013. Acute respiratory
distress syndrome: current concepts and future directions. Anaesth. Intensive
Care 41: 463–472.
44. Matute-Bello, G., G. Downey, B. B. Moore, S. D. Groshong, M. A. Matthay,
A. S. Slutsky, and W. M. Kuebler, Acute Lung Injury in Animals Study Group.
2011. An official American Thoracic Society workshop report: features and
measurements of experimental acute lung injury in animals. Am. J. Respir. Cell
Mol. Biol. 44: 725–738.
45. Morales, M. M. B., R. C. Pires-Neto, N. Inforsato, T. Lanc¸ as, L. F. F. da Silva,
P. H. N. Saldiva, T. Mauad, C. R. R. Carvalho, M. B. P. Amato, and
M. Dolhnikoff. 2011. Small airway remodeling in acute respiratory distress
syndrome: a study in autopsy lung tissue. Crit. Care 15: R4.
46. Grommes, J., and O. Soehnlein. 2011. Contribution of neutrophils to acute lung
injury. Mol. Med. 17: 293–307.
47. Williams, A. E., and R. C. Chambers. 2014. The mercurial nature of neutrophils: still
an enigma in ARDS? Am. J. Physiol. Lung Cell. Mol. Physiol. 306: L217–L230.
48. Bhatia, M., and S. Moochhala. 2004. Role of inflammatory mediators in the
pathophysiology of acute respiratory distress syndrome. J. Pathol. 202: 145–156.
49. Park, W. Y., R. B. Goodman, K. P. Steinberg, J. T. Ruzinski, F. Radella, II,
D. R. Park, J. Pugin, S. J. Skerrett, L. D. Hudson, and T. R. Martin. 2001. Cy-
tokine balance in the lungs of patients with acute respiratory distress syndrome.
Am. J. Respir. Crit. Care Med. 164: 1896–1903.
50. Baughman, R. P., K. L. Gunther, M. C. Rashkin, D. A. Keeton, and
E. N. Pattishall. 1996. Changes in the inflammatory response of the lung during
acute respiratory distress syndrome: prognostic indicators. Am. J. Respir. Crit.
Care Med. 154: 76–81.
51. Baudiß, K., C. K. Ayata, Z. Lazar, S. Cicko, J. Beckert, A. Meyer, A. Zech,
R. P. Vieira, R. Bittman, A. Go
´mez-Mun
˜oz, et al. 2015. Ceramide-1-phosphate in-
hibits cigarette smoke-induced airway inflammation. Eur. Respir. J. 45: 1669–1680.
52. Granado, M. H., P. Gangoiti, A. Ouro, L. Arana, and A. Go
´mez-Mun
˜oz. 2009.
Ceramide 1-phosphate inhibits serine palmitoyltransferase and blocks apoptosis
in alveolar macrophages. Biochim. Biophys. Acta 1791: 263–272.
53. Pettus, B. J., A. Bielawska, P. Subramanian, D. S. Wijesinghe, M. Maceyka,
C. C. Leslie, J. H. Evans, J. Freiberg, P. Roddy, Y. A. Hannun, and C. E. Chalfant.
2004. Ceramide 1-phosphate is a direct activator of cytosolic phospholipase A2. J.
Biol. Chem. 279: 11320–11326.
54. Go
´mez-Mun
˜oz, A., J. Y. Kong, B. Salh, and U. P. Steinbrecher. 2004. Ceramide-
1-phosphate blocks apoptosis through inhibition of acid sphingomyelinase in
macrophages. J. Lipid Res. 45: 99–105.
55. Park, G. Y., and J. W. Christman. 2006. Nuclear factor kappa B is a promising
therapeutic target in inflammatory lung disease. Curr. Drug Targets 7: 661–668.
56. Yang, K.-Y., J. J. Arcaroli, and E. Abraham. 2003. Early alterations in neutrophil
activation are associated with outcome in acute lung injury. Am. J. Respir. Crit.
Care Med. 167: 1567–1574.
57. Schwartz, M. D., E. E. Moore, F. A. Moore, R. Shenkar, P. Moine, J. B. Haenel,
and E. Abraham. 1996. Nuclear factor-kappa B is activated in alveolar macro-
phages from patients with acute respiratory distress syndrome. Crit. Care Med.
24: 1285–1292.
58. Hankins, J. L., K. E. Ward, S. S. Linton, B. M. Barth, R. V. Stahelin, T. E. Fox,
and M. Kester. 2013. Ceramide 1-phosphate mediates endothelial cell invasion
via the annexin a2-p11 heterotetrameric protein complex. J. Biol. Chem. 288:
19726–19738.
8 C1P REDUCES LPS-INDUCED ACUTE LUNG INJURY
at Med Universitatsklinik Bibliothek on January 29, 2016http://www.jimmunol.org/Downloaded from
... The mechanism by which neutrophils are recruited to the lungs after LPS exposure requires the production of chemokines produced by airway epithelial cells [32,33]. Based ...
... The mechanism by which neutrophils are recruited to the lungs after LPS exposure requires the production of chemokines produced by airway epithelial cells [32,33]. Based Figure 1B-D that demonstrated an increase in neutrophil recruitment, we expected that there would be an increase in the neutrophil-recruiting chemokine IL-8 (the murine equivalent of IL-8 is called the keratinocyte-derived chemokine ( KC) [34]) in LPStreated mice. ...
... In line with previous human and murine studies [12,30,32], our data indicate that Geitlerinema sp. LPS induces pro-inflammatory cytokines after intratracheal exposure. ...
Article
Full-text available
Glucocorticoid-resistant asthma, which predominates with neutrophils instead of eosinophils, is an increasing health concern. One potential source for the induction of neutrophil-predominant asthma is aerosolized lipopolysaccharide (LPS). Cyanobacteria have recently caused significant tidal blooms, and aerosolized cyanobacterial LPS has been detected near the cyanobacterial overgrowth. We hypothesized that cyanobacterial LPS contributes to lung inflammation by increasing factors that promote lung inflammation and neutrophil recruitment. To test this hypothesis, c57Bl/6 mice were exposed intranasally to LPS from the cyanobacterium member, Geitlerinema sp., in vivo to assess neutrophil infiltration and the production of pro-inflammatory cytokines and chemokines from the bronchoalveolar fluid by ELISA. Additionally, we exposed the airway epithelial cell line, A549, to Geitlerinema sp. LPS in vitro to confirm that airway epithelial cells were stimulated by this LPS to increase cytokine production and the expression of the adhesion molecule, ICAM-1. Our data demonstrate that Geitlerinema sp. LPS induces lung neutrophil infiltration, the production of pro-inflammatory cytokines such as Interleukin (IL)-6, Tumor necrosis factor-alpha, and Interferongamma as well as the chemokines IL-8 and RANTES. Additionally, we demonstrate that Geitlerinema sp. LPS directly activates airway epithelial cells to produce pro-inflammatory cytokines and the adhesion molecule, Intercellular Adhesion Molecule-1 (ICAM-1), in vitro using the airway epithelial cell line, A549. Based on our findings that use Geitlerinema sp. LPS as a model system, the data indicate that cyanobacteria LPS may contribute to the development of glucocorticoid-resistant asthma seen near water sources that contain high levels of cyanobacteria.
... The intervention has a major impact on pulmonary microcirculation, causing neutrophil infiltration of respiratory units and intra-alveolar edema entailing 'cytokine storm' and reactive oxygen species outburst, critically affecting the airway and gas exchange functionalities and highly lethal [37][38][39][40][41]. The local inflammatory reaction is triggered by LPS binding to TLR-4 at the pulmonary epithelium surface, which stimulates the NF-κB-dependent production of pro-inflammatory cyto-/chemokines including TNF-α, IL-1, IL-6 and notably MCP-1, a crucial attractant of mononuclear leukocytes to interalveola [42]. Thus, the intratracheal LPS administration provides a feasible model of human ARDS to be used for the validation of potential influence of M2-polarized macrophages on the pulmonary inflammation deployment. ...
... PCRs were set in triplicates with qPCRmix-HS SYBR master mixes (Evrogen, Russia) and target-specific primers designed using Primer-BLAST tool (NCBI, USA) and custom-ordered from Evrogen. Gene expression was quantified by threshold cycle (Ct) approach [42] against Gapdh as housekeeping reference target. The genes of interest were Arg1, Il10 and Tgfb in cultured cells and Tnfa, Nos2, Cd38, Arg1, Vegfa and Tgfb in lung tissues. ...
Article
Full-text available
Innate immunity reactions are core to any immunological process, including systemic inflammation and such extremes as acute respiratory distress syndrome (ARDS) and cytokine storm. Macrophages, the key cells of innate immunity, show high phenotypic plasticity: depending on microenvironmental cues, they can polarize into M1 (classically activated, pro-inflammatory) or M2 (alternatively activated, anti-inflammatory). The anti-inflammatory M2 macrophage polarization-based cell therapies constitute a novel prospective modality. Systemic administration of ‘educated’ macrophages is intended at their homing in lungs in order to mitigate the pro-inflammatory cytokine production and reduce the risks of ‘cytokine storm’ and related severe complications. Acute respiratory distress syndrome (ARDS) is the main mortality factor in pneumonia including SARS-CoV-associated cases. This study aimed to evaluate the influence of infusions of RAW 264.7 murine macrophage cell line polarized towards M2 phenotype on the development of LPS-induced ARDS in mouse model. The results indicate that the M2-polarized RAW 264.7 macrophage infusions in the studied model of ARDS promote relocation of lymphocytes from their depots in immune organs to the lungs. In addition, the treatment facilitates expression of M2-polarization markers Arg1, Vegfa and Tgfb and decreases of M1-polarization marker Cd38 in lung tissues, which can indicate the anti-inflammatory response activation. However, treatment of ARDS with M2-polarized macrophages didn't change the neutrophil numbers in the lungs. Moreover, the level of the Arg1 protein in lungs decreased throughtout the treatment with M2 macrophages, which is probably because of the pro-inflammatory microenvironment influence on the polarization of macrophages towards M1. Thus, the chemical polarization of macrophages is unstable and depends on the microenvironment. This adverse effect can be reduced through the use of primary autologous macrophages or some alternative methods of M2 polarization, notably siRNA-mediated.
... Currently, both pro-and anti-inflammatory properties of the phospho-sphingolipid C1P have been reported, and which results depend on the cellular compartment of production and signaling (10,(23)(24)(25)(26)(27). Here we have shown that C8-C1P but not C16-C1P treatment was able to reduce pro-inflammatory markers (CD44, CD80, and HLA-DR in addition to IL-6 secretion) in humanisolated CD14 + monocytes challenged with LPS. ...
... The synthetic sphingophospholipid, C8-C1P, has demonstrated extensive therapeutic potential, particularly in some pathological scenarios such as non-allergic asthma, smoke-induced airway inflammation, chronic diabetic wounds, ischemia, and other conditions involving injured tissues and a proinflammatory background (12). One of the more remarkable advantages is the lipidic nature of C8-C1P which contributes to its chemical stability and provides versatility for distinct possible pharmacological presentations (eg, inhalable aerosols, injectable emulsions, pills, ointments, and creams, among many others) (10,11,26,49). However, our knowledge about C1P remains limited and many questions are still unanswered regarding the mechanistic action and cellular response to this phospholipid. ...
Article
Full-text available
Monocytes (Mo) are highly plastic myeloid cells that differentiate into macrophages after extravasation, playing a pivotal role in the resolution of inflammation and regeneration of injured tissues. Wound-infiltrated monocytes/macrophages are more pro-inflammatory at early time points, while showing anti-inflammatory/pro-reparative phenotypes at later phases, with highly dynamic switching depending on the wound environment. Chronic wounds are often arrested in the inflammatory phase with hampered inflammatory/repair phenotype transition. Promoting the tissue repair program switching represents a promising strategy to revert chronic inflammatory wounds, one of the major public health loads. We found that the synthetic lipid C8-C1P primes human CD14⁺ monocytes, restraining the inflammatory activation markers (HLA-DR, CD44, and CD80) and IL-6 when challenged with LPS, and preventing apoptosis by inducing BCL-2. We also observed increased pseudo-tubule formation of human endothelial-colony-forming cells (ECFCs) when stimulated with the C1P-macrophages secretome. Moreover, C8-C1P-primed monocytes skew differentiation toward pro-resolutive-like macrophages, even in the presence of inflammatory PAMPs and DAMPs by increasing anti-inflammatory and pro-angiogenic gene expression patterns. All these results indicate that C8-C1P could restrain M1 skewing and promote the program of tissue repair and pro-angiogenic macrophage.
... When external stimuli such as inflammatory factors, cyclooxygenase (COX2), chemokines, adhesion factors, and colony-stimulating factors, bind to their corresponding receptors, NF-κB signaling is activated [32]. Studies have found that ALI development is closely correlated to the activation of NF-κB signaling, and inhibiting the NF-κB pathway is found to effecttively alleviate the symptoms of ALI [33,34]. Herein, NF-κB signaling was found markedly activated in ALI mice, in line with previous studies [35,36]. ...
Article
Full-text available
Acute lung injury (ALI) is characterized by severely damaged alveoli and blood vessels, seriously affecting the health of patients and causing a high mortality rate. The pathogenesis of ALI is complex, with inflammatory reactions and oxidative stress (OS) mainly involved. S14G humanin (HNG) is derived from humanin (HN), which is claimed with promising anti-inflammatory functions. Herein, the protective influence of HNG on ALI will be explored in a mouse model. The ALI model was established in mice via intratracheal instillation of 3 mg/kg LPS, followed by an intraperitoneal injection of 3 and 6 mg/kg HNG, respectively. Thicker alveolar walls, aggravated neutrophil infiltration, and increased wet weight/dry weight (W/D) ratio were observed in ALI mice, accompanied by an aggravated apoptotic state, all of which were notably alleviated by HNG. Furthermore, increased number of total cells and neutrophils in bronchoalveolar lavage fluid (BALF), elevated secretion of inflammatory cytokines, enhanced reactive oxygen species (ROS) and Malondialdehyde (MDA) levels, and declined superoxide dismutase-2 (SOD2) levels were observed in ALI mice, which were markedly ameliorated by HNG. Moreover, the upregulated levels of NOD-like receptor family pyrin domain containing 3 (NLRP3), caspase-1, and caspases cleave gasdermin D N/caspases cleave gasdermin D FL (GSDMD N/GSDMD FL) in ALI mice were signally repressed by HNG. Lastly, the upregulation of Toll-like receptor 4 (TLR4) and p-p65/p65, and downregulation of IκB-α observed in ALI mice were sharply reversed by HNG. Collectively, HNG alleviated the ALI in mice by inhibiting the activation of nuclear factor kappa B (NF-κB) signaling.
... In addition, CerP stimulates the release of arachidonic acid and the synthesis of prostanoids and proinflammatory cytokines, as demonstrated in an adenocarcinoma cell model 43 , and enhances immune defence by stimulating neutrophil phagocytosis 44 , mast cell degranulation 45 , and macrophage chemotaxis 46 . Finally, CerP can prevent LPS-induced injury by attenuating NFK-B activity 47 . ...
Article
Full-text available
This study aimed to determine the lipidome of water buffalo milk with intramammary infection (IMI) by non-aureus staphylococci (NAS), also defined as coagulase-negative staphylococci, using an untargeted lipidomic approach. Non-aureus Staphylococci are the most frequently isolated pathogens from dairy water buffalo milk during mastitis. A total of 17 milk samples from quarters affected by NAS-IMI were collected, and the lipidome was determined by liquid chromatography-quadrupole time-of-flight mass spectrometry. The results were compared with the lipidome determined on samples collected from 16 healthy quarters. The study identified 1934 different lipids, which were classified into 15 classes. The abundance of 72 lipids changed in NAS-IMI milk compared to healthy quarters. Significant changes occurred primarily in the class of free fatty acids. The results of this study provided first-time insight into the lipidome of dairy water buffalo milk. Moreover, the present findings provide evidence that NAS-IMI induces changes in water buffalo milk's lipidome.
... Recently, Baudiß et al. reported that C1P suppresses cigarette smoke-induced airway inflammation (15). LPS-induced lung injury is also reported to be ameliorated by C1P as it suppresses proinflammatory responses (16). These observations suggest that C1P plays a role in mitigating the inflammatory symptoms of lung disease. ...
Article
Full-text available
Idiopathic pulmonary fibrosis (IPF) is the most common idiopathic interstitial pneumonias. Lyso- phosphatidic acid (LPA) and sphingosine 1-phosphate (S1P) are signaling lipids that evoke growth factor-like responses to many cells. Recent studies revealed the involvement of LPA and S1P in the pathology of IPF. In this study, we determined LPA, S1P and ceramide 1-phosphate (C1P) in peripheral blood plasma of IPF patients, and examined correlation to the vital capacity of lung (VC), an indicator of development of fibrosis. Blood plasma samples were taken from eleven patients with IPF and seven healthy volunteers. The lipids of the sample were extracted and subjected to liquid chromatography-tandem mass spectrometry for analysis. Results showed that there is a significant negative correlation between VC and plasma LPA levels, indicating that IPF patients with advanced fibrosis had higher concentration of LPA in their plasma. Average of S1P levels were significantly high- er in IPF patients than those in healthy subjects. Although it is not statistically significant, a similar correlation trend that observed in LPA levels also found between VC and S1P levels. These results indicated that plasma LPA and S1P may be associated with deterioration of pulmonary function of IPF patients.
... These enzymes are not found in the main Cer formation pathways, named above. Of interest, C1P is highly involved in processes related to stroke such as cell migration (Granado et al., 2009b;Kim et al., 2012;Arana et al., 2013;Vera et al., 2021), invasion involving MMPs (Ordoñez et al., 2016a), cell proliferation through NO (Gangoiti et al., 2008) or vascular endothelial growth factor (VEGF) (Ouro et al., 2017), cell survival Gomez-Larrauri et al., 2021b), metabolism , and inflammation (Nakamura et al., 2006;Gomez-Muñoz et al., 2013;Baudiß et al., 2016). What makes C1P also an interesting target for the study of its involvement in stroke. ...
Article
Full-text available
Ischemic stroke, caused by the interruption of blood flow to the brain and subsequent neuronal death, represents one of the main causes of disability in worldwide. Although reperfusion therapies have shown efficacy in a limited number of patients with acute ischemic stroke, neuroprotective drugs and recovery strategies have been widely assessed, but none of them have been successful in clinical practice. Therefore, the search for new therapeutic approaches is still necessary. Sphingolipids consist of a family of lipidic molecules with both structural and cell signaling functions. Regulation of sphingolipid metabolism is crucial for cell fate and homeostasis in the body. Different works have emphasized the implication of its metabolism in different pathologies, such as diabetes, cancer, neurodegeneration, or atherosclerosis. Other studies have shown its implication in the risk of suffering a stroke and its progression. This review will highlight the implications of sphingolipid metabolism enzymes in acute ischemic stroke.
Article
Fragesin, a traditional Chinese medicine, has been shown to exert anti‐inflammatory effect. The aim of this study was to figure out the possible effectiveness of the fargesin, and to invest the mechanisms by which it works in the cadmium‐induced lung injury in mice. Fargesin was given 1 h before cadmium treatment for 7 days. Then, the bronchoalveolar lavage fluid (BALF) were harvested to test inflammatory cells and pro‐inflammatory cytokine production. Lung histopathological changes, myeloperoxidase (MPO) activity, and aryl hydrocarbon receptor (AhR) and nuclear factor kappa B (NF‐κB) activation were measured. Fargesin dose‐dependently reduced inflammatory cells and pro‐inflammatory cytokines in BALF, improved lung histopathological injury, and inhibited lung wet/dry ratio and MPO activity. Furthermore, fargesin inhibited cadmium‐induced NF‐κB activation. In addition, fargesin was found to increase AhR expression. In conclusion, fargesin attenuates cadmium‐induced lung injury may be via activating AhR, which subsequently suppressing the inflammatory response.
Article
Ceramide, a central molecule of sphingolipid metabolism, is phosphorylated to ceramide-1-phosphate (C1P) by ceramide kinase (CerK). The CerK/C1P pathway regulates many cellular functions, but its roles in immune/inflammation-related (IIR) diseases in vivo are not well known. Sepsis is an acute systemic inflammatory disease accompanied by damage/dysfunction in multiple organs. In the present study, we investigated the effects of CerK knockout on the onset/progression of sepsis-related events in lipopolysaccharide (LPS)-treated sepsis-model mice. In CerK-null mice, the lethality at 48 h after i.v. injection of LPS was significantly increased compared with that in wild-type (WT) mice. The increased lethality by CerK knockout was reproduced in mice treated with i.p. injections of LPS. Changes in serum levels of 23 IIR molecules, including cytokines and chemokines, were measured. In WT mice, levels of these molecules increased 4 and/or 20 h after i.v. injection of LPS. Although the basal levels of IIR molecules were not affected, LPS-induced increases in interleukin-17 (IL-17), C-C motif chemokine ligands (CCL-2 and CCL-11), and tumor necrosis factor-α were significantly up-regulated, whereas IL-2 levels were slightly down-regulated by CerK knockout. Putative mechanisms for the CerK/C1P pathway-mediated regulation of IIR molecules and increased lethality in LPS-treated mice are discussed.
Article
Background: Background: Acute lung injury (ALI) is one kind of frequently occurred emergency in Intensive Care Unite with a high mortality. The underlying causes are uncontrolled inflammatory reactions and intractable hypoxemia, which are difficult to control and improve. In the past 10 years, gas medical studies have found that both hydrogen molecules and oxygen molecules have protective effects on acute lung injury by improving inflammatory reactions and hypoxia, respectively. Oxygen is an oxidant and hydrogen is an antioxidant. In this study, we investigated the combined effect of above two-gas molecular on lipopolysaccharide (LPS) -induced acute lung injury. Methods: To clarify whether the combination of hydrogen and oxygen could increase or cancel out the protective effect, an ALI mice model induced by intraperitoneal injection of LPS was established, and the degree of lung tissue and mitochondria damage was evaluated based on the pathological sections, inflammatory factors, wet-dry ratio, bronchoalveolar lavage fluid (BALF). Immunohistochemistry, electron microscopy, western blotting and other detection methods also used to evaluate the therapeutic effect on acute lung injury model. Results: We observed that the combined protective effect of hydrogen and oxygen was superior to their respective protective effects, and the specific molecular mechanisms of the two therapies might be different. Conclusion: Hydrogen plays a more important role in the inflammatory and anti-apoptosis mechanisms, while oxygen improves hypoxia of the body, and thus, its molecular mechanism may be closely associated to the hypoxia pathways.
Article
Full-text available
Sphingolipids are involved in the pathogenesis of inflammatory diseases. The central molecule is ceramide, which can be converted into ceramide-1-phosphate (C1P). Although C1P can exert anti- and pro-inflammatory effects, its influence on cigarette smoke (CS)-induced lung inflammation is unknown. We aimed to clarify the role of C1P in the pathogenesis of CS-triggered pulmonary inflammation and emphysema in humans and mice. The effects of C1P were addressed on CS-induced lung inflammation in C57BL/6 mice, CS extract-triggered activation of human airway epithelial cells (AECs) and neutrophils from patients with chronic obstructive pulmonary disease. Differential cell counts in bronchoalveolar lavage fluid were determined by flow cytometry and pro-inflammatory cytokines were measured by ELISA. Expression and DNA binding of nuclear factor (NF)-κB and neutral sphingomyelinase (nSMase) were quantified by PCR, electrophoretic mobility shift and fluorometric assays. C1P reduced CS-induced acute and chronic lung inflammation and development of emphysema in mice, which was associated with a reduction in nSMase and NF-κB activity in the lungs. nSMase activity in human serum correlated negatively with forced expiratory volume in 1 s % predicted. In human AECs and neutrophils, C1P inhibited CS-induced activation of NF-κB and nSMase, and reduced pro-inflammatory cytokine release. Our results suggest that C1P is a potential target for anti-inflammatory treatment in CS-induced lung inflammation. Copyright ©ERS 2015.
Article
Full-text available
The acute respiratory distress syndrome (ARDS) is a life-threatening lung condition resulting from direct and indirect insults to the lung. It is characterised by disruption of the endothelial-epithelial barrier, alveolar damage, pulmonary oedema and respiratory failure. A key feature of ARDS is the accumulation of neutrophils in the lung microvasculature, interstitium and alveolar space. Despite a clear association between neutrophil influx into the lung and disease severity, there is some debate as to whether neutrophils directly contribute to disease pathogenesis. The primary function of neutrophils is to provide immediate host defence against pathogenic microorganisms. Neutrophils release numerous antimicrobial factors such as reactive oxygen species, proteinases and neutrophil extracellular traps (NETs). However, these factors are also toxic to host cells and can result in bystander tissue damage. The excessive accumulation of neutrophils in ARDS may therefore contribute to disease progression. Central to neutrophil recruitment is the release of chemokines, including the archetypal neutrophil chemoattractant IL-8, from resident pulmonary cells. However, the chemokine network in the inflamed lung is complex and may involve several other chemokines, including CXCL10, CCL2 and CCL7. This review will therefore focus on the experimental and clinical evidence supporting neutrophils as key players in ARDS and the chemokines involved in recruiting them into the lung.
Article
Full-text available
The purpose of this review is to examine and discuss the incidence and outcome of patients with the acute respiratory distress syndrome (ARDS). This is a challenging task, as there is no specific clinical sign or diagnostic test that accurately identifies and adequately defines this syndrome. This review will focus on published epidemiological studies reporting population-based incidence of ARDS, as defined by the American-European Consensus Conference criteria. In addition, the current outcome figures for ARDS patients reported in observational and randomized controlled trials will be reviewed. The focus will be on studies published since 2000, when the ARDSnet study on protective mechanical ventilation was published, although particular emphasis will be on those articles published in the last 24 months. On the basis of current evidence, and despite the order of magnitude of reported European and USA incidence figures, it seems that the incidence and overall mortality of ARDS has not changed substantially since the original ARDSnet study. The current mortality of adult ARDS is still greater than 40%.
Article
Full-text available
Ceramide, a product of agonist-stimulated sphingo-myelinase activation, is known to be generated during the phagocytosis of antibody-coated erythrocytes by polymorphonuclear leukocytes. Agonist-stimulated formation of ceramide-1-phosphate is now shown to occur in 32 PO 4-labeled neutrophils. Ceramide-1-phosphate is formed by a calcium-dependent ceramide kinase, found predominately in the neutrophil plasma membrane. The neutrophil kinase is specific for ceramide because, in contrast to the bacterial diglyceride kinase, ceramide is not phosphorylated under conditions specific for diglyc-eride phosphorylation. Conversely, 1,2-diacylglycerol does not serve as substrate for the neutrophil ceramide kinase. Ceramide kinase activation occurs in a time-dependent fashion, reaching peak activity 10 min after formyl peptide stimulation and challenge with anti-body-coated erythrocytes. The lipid kinase activity is optimal at pH 6.8. Because the formation of the phagoly-sosome is a critical event in phagocytosis, the effect of ceramide-1-phosphate in promoting the fusion of lipo-somes was determined. Both the addition of increasing concentrations of sphingomyelinase D and ceramide-1-phosphate promoted liposomal fusion. In summary, cer-amide-1-phosphate is formed during phagocytosis through activation of ceramide kinase. Ceramide-1-phosphate may promote phagolysosome formation.
Article
Full-text available
Acute respiratory distress syndrome is one of the leading causes of death in critically ill patients. Recent advances in supportive care have led to a moderate improvement in mortality. In particular, a much lower mortality rate than expected was evident in the severest category of patients (requiring extracorporeal membrane oxygenation) in Australia during the recent H1N1 pandemic. Though improvements in supportive care may have provided some benefit, there remains an absence of effective biological agents that are necessary to achieve further incremental reduction in mortality. This article will review the evidence available for current treatment strategies and discuss future research directions that may eventually improve outcomes in this important global disease.
Article
Full-text available
The bioactive sphingolipid, ceramide 1-phosphate (C-1-P), has been implicated as an extracellular chemotactic agent directing cellular migration in hematopoietic stem/progenitor cells and macrophages. However, interacting proteins that could mediate these actions of C-1-P have, thus far, eluded identification. We have now identified and characterized interactions between ceramide 1-phosphate and the annexin a2-p11 heterotetramer constituents. This C-1-P-receptor complex is capable of facilitating cellular invasion. Herein, we demonstrate in both coronary artery macrovascular endothelial cells and retinal microvascular endothelial cells that C-1-P induces invasion through an extracellular matrix barrier. By employing surface plasmon resonance, lipid-binding ELISA, and mass spectrometry technologies, we have demonstrated that the heterotetramer constituents bind to C-1-P. Although the annexin a2-p11 heterotetramer constituents do not bind the lipid C-1-P exclusively, other structurally similar lipids, such as phosphatidylserine, sphingosine 1-phosphate, and phosphatidic acid, could not elicit the potent chemotactic stimulation observed with C-1-P. Further, we show that siRNA-mediated knockdown of either annexin a2 or p11 protein significantly inhibits C-1-P-directed invasion, indicating that the heterotetrameric complex is required for C-1-P-mediated chemotaxis. These results imply that extracellular C-1-P, acting through the extracellular annexin a2-p11 heterotetrameric protein, can mediate vascular endothelial cell invasion. Background: Extracellular ceramide 1-phosphate is presumed to interact with extracellular proteins to mediate cellular invasion. These proteins are unidentified. Results: C-1-P interacts with both annexin a2 and p11 proteins. C-1-P-mediated vascular endothelial cell invasion requires expression of these proteins. Conclusion: Extracellular C-1-P mediates invasion via an interaction with the annexin a2-p11 heterotetramer. Significance: Gradients of C-1-P may guide vascular endothelial cell invasion during wound healing.
Article
Advances in supportive care and ventilator management for acute respiratory distress syndrome (ARDS) have resulted in declines in short-term mortality, but risks of death after survival to hospital discharge have not been well described. Our objective was to quantify the difference between short-term and long-term mortality in ARDS and to identify risk factors for death and causes of death at 1 year among hospital survivors. This multi-intensive care unit, prospective cohort included patients with ARDS enrolled between January 2006 and February 2010. We determined the clinical characteristics associated with in-hospital and 1-year mortality among hospital survivors and utilized death certificate data to identify causes of death. Of 646 patients hospitalized with ARDS, mortality at 1 year was substantially higher (41 %, 95 % CI 37-45 %) than in-hospital mortality (24 %, 95 % CI 21-27 %), P < 0.0001. Among 493 patients who survived to hospital discharge, the 110 (22 %) who died in the subsequent year were older (P < 0.001) and more likely to have been discharged to a nursing home, other hospital, or hospice compared to patients alive at 1 year (P < 0.001). Important predictors of death among hospital survivors were comorbidities present at the time of ARDS, and not living at home prior to admission. ARDS-related measures of severity of illness did not emerge as independent predictors of mortality in hospital survivors. Despite improvements in short-term ARDS outcomes, 1-year mortality is high, mostly because of the large burden of comorbidities, which are prevalent in patients with ARDS.
Article
We investigated the expression and the role of hypoxia-inducible factor 1α (HIF-1α) on the desferrioxamine (DFX)-induced cytokine production in human mast cells, HMC-1 cells. HIF-1α mRNA was constitutively expressed in mast cell lines including the P815, RBL-2H3, and HMC-1. DFX (100μM) resulted in a great increase in protein levels of HIF-1α in HMC-1 cells, but it did not affect HIF-1α mRNA expression. Iron (HIF-1 inhibitor) inhibited increase of HIF-1α and NF-κB protein levels. Pyrriolidine-dithiocarbamate (PDTC, NF-κB inhibitor) inhibited increase of NF-κB protein levels, but it slightly increased HIF-1α protein levels. In addition, DFX significantly increased the production of IL-6, IL-8, and TNF-α in HMC-1 (P